Guidance for Industry Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment

Size: px
Start display at page:

Download "Guidance for Industry Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment"

Transcription

1 Guidance for Industry Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Comments and suggestions regarding this draft document should be submitted within 60 days of publication in the Federal Register of the notice announcing the availability of the draft guidance. Submit comments to the Division of Dockets Management (HFA-305), Food and Drug Administration, 5630 Fishers Lane, rm. 1061, Rockville, MD All comments should be identified with the docket number listed in the notice of availability that publishes in the Federal Register. For questions regarding this draft document contact Patrick Guinn (CDER) at , or Miles Braun (CBER) at U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER) May 2004 Clinical Medical

2 Guidance for Industry Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment Additional copies are available from: Office of Training and Communication Division of Drug Information, HFD-240 Center for Drug Evaluation and Research Food and Drug Administration 5600 Fishers Lane Rockville, MD (Tel) or Office of Communication, Training, and Manufacturers Assistance, HFM-40 Center for Biologics Evaluation and Research Food and Drug Administration 1401 Rockville Pike, Rockville, MD (Tel) Voice Information System at or U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER) May 2004 Clinical Medical

3 TABLE OF CONTENTS I. INTRODUCTION...1 II. BACKGROUND...1 A. PDUFA III S RISK MANAGEMENT GUIDANCE GOAL...1 B. OVERVIEW OF THE RISK MANAGEMENT GUIDANCES...2 III. THE ROLE OF PHARMACOVIGILANCE IN RISK MANAGEMENT...3 IV. IDENTIFYING AND DESCRIBING SAFETY SIGNALS: FROM CASE REPORTS TO CASE SERIES...4 A. GOOD REPORTING PRACTICE...4 B. CHARACTERISTICS OF A GOOD CASE REPORT...5 C. DEVELOPING A CASE SERIES AND ASSESSING CAUSALITY OF INDIVIDUAL CASE REPORTS...6 D. SUMMARY DESCRIPTIVE ANALYSIS OF A CASE SERIES...7 E. USE OF DATA MINING TO IDENTIFY PRODUCT-EVENT COMBINATIONS...8 F. SAFETY SIGNALS THAT MAY WARRANT FURTHER INVESTIGATION...9 G. PUTTING THE SIGNAL INTO CONTEXT: CALCULATING REPORTING RATES VS. INCIDENCE RATES...10 V. BEYOND CASE REVIEW: INVESTIGATING A SIGNAL THROUGH OBSERVATIONAL STUDIES...11 A. PHARMACOEPIDEMIOLOGIC SAFETY STUDIES...12 B. REGISTRIES...14 C. SURVEYS...15 VI. INTERPRETING SAFETY SIGNALS: FROM SIGNAL TO POTENTIAL SAFETY RISK...16 VII. BEYOND ROUTINE PHARMACOVIGILANCE: DEVELOPING A PHARMACOVIGILANCE PLAN...17

4 Draft Not for Implementation Guidance for Industry 1 Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment This draft guidance, when finalized, will represent the Food and Drug Administration's (FDA's) current thinking on this topic. It does not create or confer any rights for or on any person and does not operate to bind FDA or the public. You can use an alternative approach if the approach satisfies the requirements of the applicable statutes and regulations. If you want to discuss an alternative approach, contact the FDA staff responsible for implementing this guidance. If you cannot identify the appropriate FDA staff, call the appropriate number listed on the title page of this guidance. I. INTRODUCTION This document provides guidance to industry on good pharmacovigilance practices and pharmacoepidemiologic assessment of observational data regarding drugs, including biological drug products (excluding blood and blood components). 2 Specifically, this document provides guidance on (1) safety signal identification, (2) pharmacoepidemiologic assessment and safety signal interpretation, and (3) pharmacovigilance plan development. FDA's guidance documents, including this guidance, do not establish legally enforceable responsibilities. Instead, guidances describe the Agency's current thinking on a topic and should be viewed only as recommendations, unless specific regulatory or statutory requirements are cited. The use of the word should in Agency guidances means that something is suggested or recommended, but not required. II. BACKGROUND A. PDUFA III s Risk Management Guidance Goal 1 This guidance has been prepared by the PDUFA III Pharmacovigilance Working Group, which includes members from the Center for Drug Evaluation and Research (CDER) and the Center for Biologics Evaluation and Research (CBER) at the Food and Drug Administration. 2 For ease of reference, this guidance uses the term product or drug to refer to all products (excluding blood products other than plasma derivatives) regulated by CDER and CBER. Similarly, for ease of reference, this draft guidance uses the term approval to refer to both drug approval and biologic licensure. Paperwork Reduction Act Public Burden Statement: This guidance contains information collection provisions that are subject to review by the Office of Management and Budget (OMB) under the Paperwork Reduction Act of 1995 (PRA) (44 U.S.C ). The collection(s) of information in this guidance were approved under OMB Control No (until March 31, 2005) and (until August 31, 2005).

5 On June 12, 2002, Congress reauthorized, for the second time, the Prescription Drug User Fee Act (PDUFA III). In the context of PDUFA III, FDA agreed to satisfy certain performance goals. One of those goals was to produce guidance for industry on risk management activities for drug and biological products. As an initial step towards satisfying that goal, FDA sought public comment on risk management. Specifically, FDA issued three concept papers. Each paper focused on one aspect of risk management, including (1) conducting premarketing risk assessment, (2) developing and implementing risk minimization tools, and (3) performing postmarketing pharmacovigilance and pharmacoepidemiologic assessments. In addition to receiving numerous written comments regarding the three concept papers, FDA held a public workshop on April 9 11, 2003, to discuss the concept papers. FDA considered all of the comments received in producing three draft guidance documents on risk management activities: 1. Premarketing Risk Assessment (Premarketing Guidance) 2. Development and Use of Risk Minimization Action Plans (RiskMAP Guidance) 3. Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment (Pharmacovigilance Guidance) B. Overview of the Risk Management Guidances Like the concept papers that preceded them, each of the three draft guidance documents focuses on one aspect of risk management. The Premarketing Guidance and the Pharmacovigilance Guidance focus on premarketing and postmarketing risk assessment, respectively. The RiskMAP Guidance focuses on risk minimization. Together, risk assessment and risk minimization form what FDA calls risk management. Specifically, risk management is an iterative process of (1) assessing a product s benefit-risk balance, (2) developing and implementing tools to minimize its risks while preserving its benefits, (3) evaluating tool effectiveness and reassessing the benefitrisk balance, and (4) making adjustments, as appropriate, to the risk minimization tools to further improve the benefit-risk balance. This four-part process should be continuous throughout a product s lifecycle, with the results of risk assessment informing the sponsor s decisions regarding risk minimization. When reviewing the recommendations provided in this guidance, sponsors and applicants should keep the following points in mind: Many recommendations in this guidance are not intended to be generally applicable to all products. Industry already performs risk assessment and risk minimization activities for products during development and marketing. The Federal Food, Drug, and Cosmetic Act (FDCA) and FDA implementing regulations establish requirements for routine risk assessment and risk minimization (e.g., FDCA sec. 503(b) (21 U.S.C. 353(b)), which provides for limiting drugs to prescription status; FDA regulations regarding spontaneous adverse event reporting and FDA-approved professional labeling). As a result, many of the recommendations presented here focus on situations when a product may pose an unusual type or level of risk. To the extent possible, we have specified in the text whether a recommendation is intended to apply to all products or only this subset of products. 2

6 It is of critical importance to protect patients and their privacy during the generation of safety data and the development of risk minimization action plans. During all risk assessment and risk minimization activities, sponsors must comply with applicable regulatory requirements involving human subjects research and patient privacy. 3 FDA recommends that sponsors comply with ethical principles for patient protection. To the extent possible, this guidance conforms with FDA s commitment to harmonize international definitions and standards as appropriate. The topics covered in this guidance are being discussed in a variety of international forums. We are participating in these discussions and believe that, to the extent possible, the recommendations in this guidance reflect current thinking on related issues. When planning risk assessment and risk minimization activities, sponsors should consider stakeholder input (e.g., from consumers, pharmacists, physicians, third party payers). There are points of overlap among the three guidances. We have tried to note in the text of each guidance when areas of overlap occur and when referencing one of the other guidances might be useful. III. THE ROLE OF PHARMACOVIGILANCE IN RISK MANAGEMENT Risk assessment during product development should be conducted in a thorough and rigorous manner; however, it is impossible to identify all safety concerns during clinical trials. Once a product is marketed, there is generally a large increase in the number of patients exposed, including those with co-morbid conditions and those being treated with concomitant medical products. Therefore, postmarketing safety data collection and risk assessment based on observational data are critical for evaluating and characterizing a product's risk profile and for making informed decisions on risk minimization. In discussing postmarketing risk assessment, this guidance uses the term pharmacovigilance to mean all observational (nonrandomized) postapproval scientific and data gathering activities relating to the detection, assessment, and understanding of adverse events. This includes the use 3 See 45 CFR part 46 and 21 CFR parts 50 and 56. See also the Health Insurance Portability and Accountability Act of 1996 (HIPAA) (Public Law ) and the Standards for Privacy of Individually Identifiable Health Information (the Privacy Rule) (45 CFR part 160 and subparts A and E of part 164). The Privacy Rule specifically permits covered entities to report adverse events and other information related to the quality, effectiveness, and safety of FDA-regulated products both to manufacturers and directly to FDA (45 CFR (b)(1)(i) and (iii), and 45 CFR (a)(1)). For additional guidance on patient privacy protection, see 3

7 of pharmacoepidemiologic safety studies. These activities are undertaken with the goal of identifying and preventing these events to the extent possible. Pharmacovigilance principally involves the identification and evaluation of safety signals in reports suggesting an excess, compared to what would be expected, of adverse events associated with a product's use. Concerns about possible adverse events can, of course, arise from other sources, such as preclinical data and events associated with other products in the same pharmacologic class. Occasionally, even a single well-documented case report can be viewed as a signal, particularly if the report describes a positive rechallenge or if the event is extremely rare in the absence of drug use. Such signals generally indicate the need for further investigation, which may or may not lead to the conclusion that the product is related to the risk. After a signal is identified, it can be further assessed in terms of its magnitude, the specific populations involved, biologic plausibility, and other factors to determine whether it represents a potential safety risk and whether action should be taken. IV. IDENTIFYING AND DESCRIBING SAFETY SIGNALS: FROM CASE REPORTS TO CASE SERIES Good pharmacovigilance practice is generally based on acquiring complete data from spontaneous adverse event reports, also known as case reports. The reports are used to develop case series for interpretation. A. Good Reporting Practice Spontaneous case reports of adverse events submitted to the sponsor and FDA, and reports from other sources, such as the medical literature or clinical studies, are potential signals of adverse effects of drugs. The quality of the reports is critical for appropriate evaluation of the relationship between the product and adverse events. FDA recommends that sponsors make every attempt to obtain complete information during initial contacts and subsequent follow-up, 4 and encourages sponsors to use trained health care practitioners to query the initial reporters. FDA suggests that the queries be focused on clinically relevant information associated with the product and the adverse event. Computer-assisted interview technology or other methods developed to target specific events can help focus the line of questioning. When the report is from a consumer, it is often important to obtain permission to contact the health care practitioner familiar with the patient s adverse event to obtain further medical information and to retrieve relevant medical records, as needed. FDA suggests that the intensity and method of case follow-up be driven by the seriousness of the event reported, the report's origin (e.g., health care practitioner, patient, literature), and other factors. FDA recommends that the most aggressive follow-up efforts be directed towards serious 4 Good reporting practices are extensively addressed in a proposed FDA regulation and guidance documents. See (1) Safety Reporting Requirements for Human Drug and Biological Products, Proposed Rule, 68 Fed. Reg (March 14, 2003), (2) FDA guidance for industry on Postmarketing Reporting of Adverse Experiences, (3) FDA guidance for industry on E2C Clinical Safety Data Management: Periodic Safety Update Report (PSUR), (4) FDA guidance for industry on Postmarketing Adverse Experience Reporting for Human Drug and Licensed Biological Products: Clarification of What to Report. 4

8 adverse event reports, especially of adverse events not known to occur with the drug and that lack clinical information and other details important for case assessment. B. Characteristics of a Good Case Report Good case reports include the following elements: 1. Description of the adverse events or disease experience, including time to onset of signs or symptoms; 2. Suspected and concomitant product therapy details (i.e., dose, schedule, dates, duration); 3. Patient characteristics, including demographic information (e.g., age, race, sex), baseline medical condition prior to product therapy, co-morbid (explain in a parenthetical) conditions, use of concomitant medications, relevant family history of disease, and presence of other risk factors; 4. Documentation of the diagnosis of the events, including methods used to make the diagnosis; 5. Clinical course of the event and patient outcomes (e.g., hospitalization or death); 5 6. Therapeutic measures and laboratory data at baseline, during therapy, and subsequent to therapy, including blood levels, as appropriate; 7. Information about response to dechallenge and rechallenge; and 8. Any other relevant information (e.g., other details relating to the event or information on benefits received by the patient, if important to the assessment of the event). For reports of medication errors, good case reports also include full descriptions of the following: 1. Products involved (including the trade and established name, manufacturer, dosage form, strength, concentration, and type and size of container); 2. Sequence of events leading up to the error; 3. Work environment in which the error occurred; and 4. Types of personnel involved with the error, type of error, causes, and contributing factors. 5 Patient outcomes may not be available at the time of initial reporting. In these cases, follow-up reports can convey important information about the course of the event and serious outcomes, such as hospitalization or death. 5

9 FDA recommends that sponsors capture in the case narrative all appropriate data elements outlined in the National Coordinating Council for Medication Error Reporting and Prevention (NCC MERP) Taxonomy. 6 The taxonomy is a tool designed to categorize and analyze reports of medication errors. It provides a standard language and structure for medication error-related data collected through reports. C. Developing a Case Series and Assessing Causality of Individual Case Reports FDA suggests that sponsors initially evaluate a signal generated from postmarketing spontaneous reports through a careful review of the cases and a search for additional cases. Additional cases could be identified from the sponsor s global adverse event databases, the published literature, and other available databases, such as FDA s Adverse Event Reporting System (AERS) or Vaccine Adverse Events Reporting System (VAERS), using thorough database search strategies based on updated coding terminology (e.g., the Medical Dictionary for Regulatory Activities (or MedDRA)). Where these are available, FDA recommends that case definitions (i.e., formal criteria for including or excluding a case) be used to assess cases. In general, FDA suggests that case-level review occur before other investigations or analyses. FDA recommends that emphasis usually be placed on review of serious, unlabeled adverse events, although other events may warrant further investigation (see section IV.F. for more details). As part of the case-level review, FDA suggests that sponsors evaluate individual case reports for clinical content and completeness and follow up with reporters, as necessary. It is important to remove any duplicate reports. In assessing case reports, FDA recommends sponsors look for features that may suggest a causal relationship between the use of a product and the adverse event, including: 1. Occurrence of the adverse event in the expected time (e.g., type 1 allergic reactions occurring within days of therapy, cancers developing after years of therapy); 2. Absence of symptoms related to the event prior to exposure; 3. Evidence of positive dechallenge or positive rechallenge; 4. Consistency of the event with the established pharmacological/toxicological effects of the product, or for vaccines, consistency with established immunologic mechanisms of injury; 5. Consistency of the event with the known effects of other products in the class; 6. Existence of other supporting evidence from preclinical studies, clinical trials, and/or pharmacoepidemiologic safety studies; and 6 See for the definition of a medication error and taxonomy of medication errors. 6

10 Absence of confounding (i.e. alternative explanations for the event, such as there were no concomitant medications that could contribute to the event, there were no co- or preexisting medical conditions). FDA recommends that sponsors carefully evaluate confounded cases and should not simply dismiss them. Confounded cases are common, especially among patients with complicated medical conditions, and could still represent adverse effects of the product under review. It is important to note that apparent lack of confounding could be due to incomplete data acquisition. For any individual case report, it is rarely possible to know with a high level of certainty whether the event was caused by the product. To date, there are no internationally agreed upon standards or criteria for assessing causality in individual cases, especially for events that often occur spontaneously (e.g. stroke, pulmonary embolism). Rigorous pharmacoepidemiologic studies, such as case-control studies and cohort studies with long-term follow-up, are usually needed to assess causality in such instances. FDA does not recommend any specific categorization of causality, but the categories probable, possible, or unlikely have been used. The World Health Organization uses the following categories: 7 certain; probably/likely; possible; unlikely; conditional/unclassified; and unassessable/unclassifiable. Although FDA does not advocate a particular categorization system, if a causality assessment is undertaken, FDA suggests that the causal categories are specified. If the safety signal relates to a medication error, FDA recommends that sponsors report the root causal factors that led to the event. A number of references describing root cause analysis are available. 8 FDA recommends that sponsors follow up to the extent possible with reporters to capture a complete account of the event, focusing on the medication use systems (e.g., prescribing/order process, dispensing process, administration process), as opposed to individuals. FDA suggests that sponsors seek to identify possible failure points in the medication use system that may be informative in developing strategies to minimize future errors. D. Summary Descriptive Analysis of a Case Series After individual cases are assessed for causality, one or more of the cases may suggest a safety signal warranting additional investigation. In that event, FDA recommends that a case series be 7 World Health Organization, the Uppsala Monitoring Center, 2000, Safety Monitoring of Medicinal Products. 8 See Cohen MR (ed), 1999, Medication Errors, American Pharmaceutical Association, Washington DC; Cousins DD (ed), 1998, Medication Use: A Systems Approach to Reducing Errors, Joint Commission on Accreditation of Healthcare Organizations, Oakbrook Terrace, IL. 7

11 assembled and descriptive clinical information summarized to characterize the potential safety risk and, if possible, to identify risk factors. A case series commonly includes an analysis of the following: 1. The clinical and laboratory manifestations and course of the event; 2. Demographic characteristics of patients with events (e.g., age, gender, race); 3. Exposure duration; 4. Time from initiation of product exposure to the adverse event; 5. Doses used in cases, including labeled doses, greater than labeled doses, and overdoses; 6. Use of concomitant medications; 7. The presence of co-morbid conditions, particularly those known to cause the adverse event, such as underlying hepatic or renal impairment; 8. The route of administration (e.g., oral vs. parenteral) and lots used in patients with events; and 9. Changes in event reporting rate over calendar time or product life cycle. E. Use of Data Mining to Identify Product-Event Combinations At various stages of risk identification and assessment, looking systematically into the data by using statistical or mathematical tools, or so-called data mining, can provide additional information about the existence or characteristics of a signal. By applying data mining techniques to large adverse event databases, such as FDA s AERS or VAERS, a sponsor may be able to identify unusual or unexpected product-event combinations warranting further investigations. Data mining is not the only technique used to make causal attributions between products and adverse events. A method of data mining currently in use is the comparison of the fraction of all events reported for a particular product (e.g., liver failure), the "observed rate," with the fraction of reports for all drugs that are for that same event, the "expected rate. This analysis can be corrected for such characteristics as reporting year, age, and gender, and it is also possible to do the analysis for drugs of a specific class or for drugs that are used to treat a particular disease. The statistic (or score) used to quantify the disproportionality between the observed and expected values for a given product-event combination is compared to a threshold that is chosen by the analyst to optimize sensitivity and specificity. A signal is operationally defined as any productevent combination with a score exceeding the specified threshold. It is not unusual for a product to have several signals identified using these methods. The lower the threshold, the more likely 8

12 it is that signals of true effects will be detected, but these lower thresholds will also result in more false positive signals. Several data mining methods have been described and are worth considering, such as the Multi- Item Gamma Poisson Shrinker (MGPS) algorithm, the proportional reporting ratio (PRR) method and the Bayesian neural network approach. Except when the observed number of events is small (e.g., less than 20), these methods will generally give similar scores. These approaches are inherently exploratory and may provide insights into the patterns of adverse events particular to a given product relative to other products in the same class or to all other products. FDA recommends exercising caution when making such comparisons, however, because voluntary adverse event reporting systems such as AERS or VAERS are subject to a variety of reporting biases, because some observations could reflect concomitant treatment, not the product itself, and because the disease being treated may cause the events. Specifically, AERS or VAERS data may be affected by the submission of incomplete or duplicate reports, under-reporting, or reporting stimulated by publicity or litigation. As reporting biases may differ by product and change over time, and could change differently for different events, it is not possible to predict their impact on data mining scores. FDA recommends considering signals identified by scores that exceed a specified threshold as hypothesisgenerating. Further investigation of a product-event combination may be warranted, especially if the event is serious and unlabeled or raises other safety concerns as described in section IV.F. When data mining results are submitted to FDA, FDA suggests that they be accompanied by a careful assessment of individual case reports and any other relevant safety information, such as results from preclinical, clinical, pharmacoepidemiologic, or other available studies. F. Safety Signals That May Warrant Further Investigation FDA believes that the methods described above will permit a sponsor to identify and preliminarily characterize safety signals. The actual risk to patients cannot be known from these data because it is not possible to characterize all cases definitively and because there is invariably under-reporting of some extent and incomplete information about duration of therapy, numbers treated, etc. Safety signals that typically warrant further investigation may include, but are not limited to, the following: 1. New unlabeled adverse events, especially if serious; 2. An apparent increase in the severity of a labeled event; 3. More than a small number of serious events thought to be extremely rare; 4. New product-product, product-food, or product-dietary supplement interactions; 5. Identification of a previously unrecognized at-risk population (e.g., populations with specific racial or genetic predispositions or co-morbidities); 6. Actual or potential confusion about a product's name, labeling, packaging, or use; 9

13 Concerns arising from the way a product is used (e.g., adverse events seen at higher than labeled doses or in populations not recommended for treatment); 8. Concerns arising from potential inadequacies of a currently implemented risk minimization action plan (e.g., reports of serious adverse events that appear to reflect failure of a RiskMAP goal); and 9 9. Other concerns identified by the sponsor or FDA. G. Putting the Signal into Context: Calculating Reporting Rates vs. Incidence Rates If a sponsor determines that a safety signal warrants further investigation and analysis, it is important to put the signal, or the excess of events, into context. For this reason, calculations of the rate at which new cases of adverse events occur in the product-exposed population (i.e., the incidence rate) are the hallmark of pharmacoepidemiologic risk assessment. In pharmacoepidemiologic safety studies (see section V.A), the numerator (number of new cases) and denominator (number of exposed patients and time of exposure) may be readily ascertainable. In contrast, for spontaneously reported events, it is not possible to identify all cases because of under-reporting, and the size of the exposed population is at best an estimate. Limitations in national denominator estimates arise because: 1. National estimates of the number of patients exposed to a medical product and their duration of exposure may not be available; 2. It may be difficult to exclude patients who are not at risk for an event because their exposure is too brief or their dose is too low; and A product may be used in different populations for different indications, but use estimates are not available for the population of interest. Although we recognize these limitations, we recommend that sponsors calculate crude adverse event reporting rates as a valuable step in the investigation and assessment of adverse events. FDA suggests that sponsors calculate reporting rates by using the total number of spontaneously reported cases in the United States in the numerator and estimates of national patient exposure to product in the denominator. 11 FDA recommends that whenever possible, the number of patients exposed to the product nationwide be the estimated denominator for a reporting rate. FDA suggests that other surrogates for exposure, such as numbers of prescriptions or kilograms of product sold, only be used when patient-level estimates are unavailable. 9 For a detailed discussion of risk minimization action plan evaluation, please consult the RiskMAP Guidance. 10 See Current Challenges in Pharmacovigilance: Pragmatic Approaches, Report of the Council for International Organizations of Medical Sciences (CIOMS) Working Group V, Geneva See Rodriguez EM, Staffa JA, Graham DJ, (2001), The role of databases in drug postmarketing surveillance, Pharmacoepidemiology and Drug Safety, 10:

14 Comparisons of reporting rates can be valuable, particularly across similar products or across different product classes prescribed for the same indication. However, such comparisons are subject to substantial limitations in interpretation because of the inherent uncertainties in the numerator and denominator used. As a result, FDA suggests that a comparison of two or more reporting rates be viewed with caution and generally considered exploratory or hypothesisgenerating. Reporting rates can by no means be considered incidence rates, for either absolute or comparative purposes. To provide further context for incidence rates or reporting rates, it is helpful to have an estimate of the background rate of occurrence for the event being evaluated in the general population or, ideally, in a subpopulation with characteristics similar to that of the exposed population (e.g., premenopausal women, diabetics). These background rates can be derived from: (1) national health statistics, (2) published medical literature, or (3) ad hoc studies, particularly of subpopulations, using large automated databases or ongoing epidemiologic investigations with primary data collection. FDA suggests that comparisons of incidence rates or reporting rates to background rate estimates (that estimate representing the rate for an exposure period similar to that of the product) take into account potential differences in the data sources used to derive the incidence rates or reporting rates compared to those used to derive the background rate. While the extent of under-reporting is unknown, it is usually assumed to be substantial and may vary according to the type of product, seriousness of the event, population using the product, and other factors. As a result, a high reporting rate compared to the background rate may, in some cases, be a strong indicator that the true incidence rate is sufficiently high to be of concern. However, many other factors affect the reporting of product-related adverse events (e.g., publicity, newness of product to the market) and these factors should be considered when interpreting a high reporting rate. Also, because of under-reporting, the fact that a reporting rate is less than the background rate does not necessarily show that the product is not associated with an increased risk of an adverse event. V. BEYOND CASE REVIEW: INVESTIGATING A SIGNAL THROUGH OBSERVATIONAL STUDIES Signals warranting additional investigation can be further evaluated through carefully designed observational studies of the product s use in the real world. Such studies could include: (1) pharmacoepidemiologic safety studies, (2) registries, and (3) surveys. Although this document focuses on these three types of observational studies, there are a variety of other methods for investigating a safety signal. For example, the Premarketing Guidance discusses the large simple safety study (LSSS), which is a risk assessment method that could be used either pre- or post-approval. By focusing this guidance on certain risk assessment methods, we do not intend to advocate the use of these approaches over others. FDA encourages sponsors to consider all methods to evaluate a particular safety signal. FDA recommends that sponsors choose the method best suited to the particular signal and research question of interest. Sponsors planning to evaluate a safety signal are encouraged to communicate with FDA as their plans progress. 11

15 A. Pharmacoepidemiologic Safety Studies Pharmacoepidemiologic safety studies are nonrandomized observational studies of patients in the "real world" being treated with a particular product. The studies can be of various designs, including cohort (prospective or retrospective), case-control, nested case-control, case-crossover, or other models. 12 The results of such studies may be used to characterize one or more safety signals associated with a product. Unlike a case series, a pharmacoepidemiologic safety study has a protocol and control group and tests prespecified hypotheses. Pharmacoepidemiologic safety studies allow for the estimation of the relative risk of an outcome associated with a product, and some (e.g., cohort studies) can also provide estimates of risk (incidence) for an adverse event. Sponsors can initiate pharmacoepidemiologic safety studies at any time. They are sometimes started at the time of initial marketing, based on questions that remain after review of the premarketing data. More often, however, they are initiated when a safety signal has been identified after approval. Finally, there may also be rare occasions when a pharmacoepidemiologic safety study is initiated prior to marketing (e.g., to study the natural history of disease or patterns of product use, or to estimate background rates for adverse events). For uncommon or delayed adverse events, pharmacoepidemiologic safety studies are often the only practical choice for evaluation. Clinical trials are impractical in almost all cases when the event rates of concern are less common than 1: It may also be difficult to use clinical trials: (1) to evaluate a safety signal associated with chronic exposure to a product, exposure in populations with co-morbid conditions, or taking multiple concomitant medications, or (2) to identify certain risk factors for a particular adverse event. On the other hand, for evaluation of more common events, where the main difficulty is that they are seen relatively often in untreated patients, clinical trials are preferable to observational studies. Because pharmacoepidemiologic safety studies are observational in nature, they are more subject to confounding, effect modification, and other bias, which may make results of these types of studies more difficult to interpret than the results of clinical trials. This problem can usually be surmounted when the relative risk of exposed patients is high or the study is sufficiently large to detect small differences in relative risk. Because different products pose different benefit-risk considerations (e.g., seriousness of the disease being treated, nature and frequency of the safety signal under evaluation), it is impossible to delineate a universal set of criteria for the point at which a pharmacoepidemiologic safety study should be initiated, and the decision should be made on a case-by-case basis. When an important adverse event product association leads to questions on the product s benefit-risk balance, FDA recommends that sponsors consider whether the particular signal should be addressed with one or more pharmacoepidemiologic safety studies. If a sponsor determines that a pharmacoepidemiologic safety study is the best method for evaluating a particular signal, the design and size of the proposed study would depend on the objectives of the study and the expected frequency of the events of interest. 12 Guidelines for Good Epidemiology Practices for Drug, Device and Vaccine Research in the United States, International Society for Pharmacoepidemiology, 1996 ( 12

16 When performing a pharmacoepidemiologic safety study, FDA suggests that investigators seek to minimize bias and to account for possible confounding. Confounding by indication is one example of an important concern in performing a pharmacoepidemiologic safety study. 13 Because of the effects of bias, confounding, or effect modification, pharmacoepidemiologic studies evaluating the same hypothesis may provide different or even conflicting results. It is almost always prudent to conduct more than one study, in more than one environment and even using different designs. Agreement of the results from more than one study helps to provide reassurance that the observed results are robust. There are a number of references describing methodologies for pharmacoepidemiologic safety studies, discussing their strengths and limitations, 14 and providing guidelines to facilitate the conduct, interpretation, and documentation of such studies. 15 Consequently, this guidance document does not comprehensively address these topics. However, protocols for a pharmacoepidemiologic safety study protocol generally include: 1. Clearly specified study objectives; 2. A critical review of the literature; and 3. A detailed description of the research methods, including: the population to be studied; the data sources to be used; the projected study size and statistical power calculations; and the methods for data collection, management, and analysis. Depending on the type of pharmacoepidemiologic safety study planned, there are a variety of data sources that may be used, ranging from the prospective collection of data to the use of existing data, such as data from previously conducted clinical trials or large databases. In recent years, a number of pharmacoepidemiologic safety studies have been conducted in automated claims databases (e.g., HMO, Medicaid) that allow retrieval of records on product exposure and patient outcomes. Depending on study objectives, factors that may affect the choice of databases selected include the following: 1. Demographic characteristics of patients enrolled in the health plans (e.g., age, geographic location); 2. Turnover rate of patients in the health plans; 3. Plan coverage of the medications of interest; 4. Size of the exposed population available for study; 13 See Strom BL (ed), 2000, Pharmacoepidemiology, 3 rd edition, Chichester: John Wiley and Sons, Ltd; Hartzema AG, Porta M, and Tilson HH (eds), 1998, Pharmacoepidemiology: An Introduction, 3 rd edition, Cincinnati, OH: Harvey Whitney Books. 14 Id. 15 Guidelines for Good Epidemiology Practices for Drug, Device and Vaccine Research in the United States, International Society for Pharmacoepidemiology, 1996 ( 13

17 Availability of the outcomes of interest; 6. Ability to identify outcomes of interest using standard coding systems (e.g., International Classification of Diseases (ICD-9)); and 7. Access to medical records. Validation of diagnostic findings in claims database studies through detailed review of at least a sample of medical records is highly recommended for most pharmacoepidemiologic safety studies. If the validation of the specific outcome of interest using the proposed database has been previously reported, FDA recommends that the literature supporting the validity of the proposed study be submitted for review. FDA encourages sponsors to communicate with the Agency when pharmacoepidemiologic safety studies are being developed. B. Registries The term registry as used in pharmacovigilance and pharmacoepidemiology can have varied meanings. In this guidance document, a registry is an organized system for the collection, storage, retrieval, analysis, and dissemination of information on individual persons exposed to a specific medical intervention who have either a particular disease, a condition (e.g., a risk factor) that predisposes [them] to the occurrence of a health-related event, or prior exposure to substances (or circumstances) known or suspected to cause adverse health effects. 16 Through the creation of registries, a sponsor can follow up on safety signals identified from spontaneous case reports, literature reports, or other sources, and evaluate factors that affect the risk of adverse outcomes, such as dose, timing of exposure, or patient characteristics. 17 Registries can be particularly useful for: 1. Collecting outcome information not available in large automated databases; and 2. Collecting information from multiple sources (e.g., physician records, hospital summaries, pathology reports, vital statistics). A sponsor can initiate a registry at any time. It may be appropriate to initiate the registry at the time of initial marketing, when a new indication is approved, or when there is a need to evaluate safety signals identified from spontaneous case reports. In deciding whether to establish a registry, FDA recommends that a sponsor consider the following factors: 1. The types of additional risk information desired; 16 See Frequently Asked Questions About Medical and Public Health Registries, The National Committee on Vital and Health Statistics, at 17 FDA guidance for industry on Establishing Pregnancy Exposure Registries, August

18 The attainability of that information through other methods; and 3. The feasibility of establishing the registry. FDA recommends that sponsors electing to initiate a registry develop written protocols that provide: (1) objectives for the registry, (2) a review of the literature, and (3) a summary of relevant animal and human data. FDA suggests that protocols also contain detailed descriptions of: (1) plans for patient recruitment and follow-up, (2) methods for data collection, management, and analysis, and (3) conditions under which the registry will be terminated. A registry-based monitoring system should include carefully designed data collection forms to ensure data quality, integrity, and validation of registry findings against a sample of medical records or through interviews with health care providers. FDA recommends that the size of the registry and the period during which data will be collected be consistent with the safety questions under study and we encourage discussion with FDA prior to initiation by the sponsor. C. Surveys Patient or health care provider surveys can gather information to assess: 1. A safety signal; 2. Knowledge about labeled adverse events; 3. Use of a product as labeled, particularly when the indicated use is for a restricted population or numerous contraindications exist; 4. Compliance with the elements of a RiskMAP (e.g., whether or not a Medication Guide was provided at the time of product dispensing); and Confusion in the practicing community over sound-alike or look-alike trade names. Like a registry, a survey can be initiated by a sponsor at any time. It can be conducted at the time of initial marketing (i.e., to fulfill a postmarketing commitment) or when there is a desire to evaluate safety signals identified from spontaneous case reports. FDA suggests that sponsors electing to initiate a survey develop a written protocol that provides objectives for the survey and a detailed description of the research methods, including: (1) patient or provider recruitment and follow-up, (2) projected sample size, and (3) methods for data collection, management, and analysis. 19 FDA recommends that a survey-based monitoring system include carefully designed survey instruments and validation of survey findings against a sample of medical or pharmacy records or through interviews with health care providers. FDA recommends that survey instruments be validated or piloted before implementation. FDA suggests that sponsors consider whether survey translation and cultural validation would be important. 18 For a detailed discussion of RiskMAP evaluation, please consult the RiskMAP Guidance. 19 See 21 CFR parts 50 and 56 for FDA's regulations governing the protection of human subjects. 15

19 Sponsors are encouraged to discuss their survey development plans with FDA. VI. INTERPRETING SAFETY SIGNALS: FROM SIGNAL TO POTENTIAL SAFETY RISK After identifying a safety signal, FDA recommends that a sponsor conduct a careful case level review, assess product relatedness at the case level, and summarize the resulting case series descriptively. To help further characterize a safety signal, a sponsor can also: (1) employ data mining techniques, and (2) calculate reporting rates for comparison to background rates. Based on these findings and other available data (e.g., from preclinical or other sources), FDA suggests that a sponsor consider further study (e.g., observational studies) to establish whether or not a potential safety risk exists. When a safety signal is identified that may represent a potential safety risk, FDA recommends that a sponsor submit a synthesis of all available safety information and analyses performed, ranging from preclinical findings to current observations. In its submission to FDA, FDA requests that a sponsor present an assessment of the likelihood that the product caused the adverse event, based on available data. In contrast to causality assessment at the individual case level (discussed in section IV.C above), it may be possible to assess the degree of causality between use of a product and an adverse event when a sponsor gathers and evaluates all available safety data, including the following: 1. Spontaneously reported and published case reports; 2. Relative risks or odds ratios derived from pharmacoepidemiologic safety studies; 3. Biologic effects observed in preclinical studies and pharmacokinetic or pharmacodynamic effects; 4. Safety findings from controlled clinical trials; and 5. General marketing experience with similar products in the class. After the available safety information is presented and interpreted, FDA suggests that the submission: (1) provide an assessment of the benefit-risk balance of the product for the population of users as a whole and for identified at-risk patient populations, (2) propose steps to further investigate the signal through additional studies, and (3) propose risk minimization actions, if appropriate. 20 FDA will make its own assessment of the potential safety risk posed by the signal in question, taking into account the information provided by the sponsor and any additional relevant information known to FDA (e.g., information on other products in the same class). Factors that are typically considered include: 20 In the vast majority of cases, risk minimization will involve risk communication by incorporating appropriate language into the product s labeling. In rare instances, however, a sponsor may consider implementing a RiskMAP. Please refer to the RiskMAP Guidance for a complete discussion of RiskMAP development. 16

Guidance for Industry

Guidance for Industry Guidance for Industry Good Pharmacovigilance Practices and Pharmacoepidemiologic Assessment U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research

More information

Guidance for Industry Development and Use of Risk Minimization Action Plans

Guidance for Industry Development and Use of Risk Minimization Action Plans Guidance for Industry Development and Use of Risk Minimization Action Plans DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Comments and suggestions regarding this

More information

1201 Maryland Avenue SW, Suite 900, Washington, DC ,

1201 Maryland Avenue SW, Suite 900, Washington, DC , 1201 Maryland Avenue SW, Suite 900, Washington, DC 20024 202-962-9200, www.bio.org June 6, 2008 Dockets Management Branch (HFA-305) Food and Drug Administration 5600 Fishers Lane, Rm. 1061 Rockville, MD

More information

Draft Guidance for Industry Development and Use of Risk Minimization Action Plans

Draft Guidance for Industry Development and Use of Risk Minimization Action Plans Draft Guidance for Industry Development and Use of Risk Minimization Action Plans Docket Number [2004D-0188] Submitted to the U.S. Department of Health and Human Services Food and Drug Administration Center

More information

October 25, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Room 1061 Rockville, MD 20852

October 25, Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Room 1061 Rockville, MD 20852 701 Pennsylvania Avenue, NW Suite 800 Washington, D.C. 20004 2654 Tel: 202 783 8700 Fax: 202 783 8750 www.advamed.org Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers

More information

1201 Maryland Avenue SW, Suite 900, Washington, DC ,

1201 Maryland Avenue SW, Suite 900, Washington, DC , 1201 Maryland Avenue SW, Suite 900, Washington, DC 20024 202-962-9200, www.bio.org June 19, 2008 Dockets Management Branch (HFA-305) Food and Drug Administration 5600 Fishers Lane, Rm. 1061 Rockville,

More information

Sonja Brajovic Medical Officer, Office of Surveillance and Epidemiology Center for Drug Evaluation and Research Food and Drug Administration

Sonja Brajovic Medical Officer, Office of Surveillance and Epidemiology Center for Drug Evaluation and Research Food and Drug Administration FDA Perspective on MedDRA Coding Quality in Post marketing Safety Reports European Informal MedDRA User Group Webinar Sonja Brajovic Medical Officer, Office of Surveillance and Epidemiology Center for

More information

DRAFT GUIDANCE. This guidance document is being distributed for comment purposes only. Document issued on: July 14, 2011

DRAFT GUIDANCE. This guidance document is being distributed for comment purposes only. Document issued on: July 14, 2011 Draft Guidance for Industry and Food and Drug Administration Staff In Vitro Companion Diagnostic Devices DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Document issued

More information

Signal Detection Quantitative Analysis of Safety Data. Dr. Yusuf Tanrikulu Basel, 29th November 2016

Signal Detection Quantitative Analysis of Safety Data. Dr. Yusuf Tanrikulu Basel, 29th November 2016 Signal Detection Quantitative Analysis of Safety Data Dr. Yusuf Tanrikulu Basel, 29th November 2016 Contents Regulatory background Goals of signal detection Data sources Methods Recent developments 2 Regulatory

More information

Guidance for Industry

Guidance for Industry Guidance for Industry Cooperative Manufacturing Arrangements for Licensed Biologics Additional copies of this guidance are available from the Office of Communication, Training and Manufacturers Assistance

More information

Guidance for Industry Postmarketing Studies and Clinical Trials Implementation of Section 505(o) of the Federal Food, Drug, and Cosmetic Act

Guidance for Industry Postmarketing Studies and Clinical Trials Implementation of Section 505(o) of the Federal Food, Drug, and Cosmetic Act Guidance for Industry Postmarketing Studies and Clinical Trials Implementation of Section 505(o) of the Federal Food, Drug, and Cosmetic Act DRAFT GUIDANCE This guidance document is being distributed for

More information

Distinguishing Medical Device Recalls from Product Enhancements and Associated Reporting Requirements

Distinguishing Medical Device Recalls from Product Enhancements and Associated Reporting Requirements 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 Distinguishing Medical Device Recalls from Product Enhancements and Associated Reporting Requirements Draft Guidance for Industry

More information

Guidance for Industry and FDA Staff Procedures for Handling Post-Approval Studies Imposed by PMA Order

Guidance for Industry and FDA Staff Procedures for Handling Post-Approval Studies Imposed by PMA Order Guidance for Industry and FDA Staff Procedures for Handling Post-Approval Studies Imposed by PMA Order Document issued on: [Level 2, June 15, 2009] This guidance supersedes the document issued under this

More information

Chin Koerner Executive Director US Regulatory and Development Policy

Chin Koerner Executive Director US Regulatory and Development Policy Chin Koerner Executive Director US Regulatory and Development Policy Novartis Pharmaceuticals Corporation 1700 Rockville Pike Suite 510 Rockville, MD 20852 Tel 301.468.5607 Fax 301.468.5614 Email: Chin.Koerner@novartis.com

More information

Guidance for Industry, Review Staff, and the Clinical Community

Guidance for Industry, Review Staff, and the Clinical Community Guidance for Industry, Review Staff, and the Clinical Community Guidance on Criteria and Approaches for Postmarket Surveillance Document Issued on: November 2, 1998 U.S. Department of Health and Human

More information

Guidance for Industry

Guidance for Industry Guidance for Industry Cooperative Manufacturing Arrangements for Licensed Biologics DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Comments and suggestions regarding

More information

PHARMA CONGRESS. Pharmacovigilance and Drug Safety: Assessing Future Regulatory and Compliance Developments. October 28, 2008

PHARMA CONGRESS. Pharmacovigilance and Drug Safety: Assessing Future Regulatory and Compliance Developments. October 28, 2008 PHARMA CONGRESS October 28, 2008 Pharmacovigilance and Drug Safety: Assessing Future Regulatory and Compliance Developments Beverly H. Lorell, MD Senior Medical & Policy Advisor King & Spalding LLP Assessing

More information

Quality check of spontaneous adverse drug reaction reporting forms of different countries y

Quality check of spontaneous adverse drug reaction reporting forms of different countries y pharmacoepidemiology and drug safety (2010) Published online in Wiley Online Library (wileyonlinelibrary.com).2004 ORIGINAL REPORT Quality check of spontaneous adverse drug reaction reporting forms of

More information

Pharmacovigilance & Signal Detection

Pharmacovigilance & Signal Detection Pharmacovigilance & Signal Detection 30 th International Conference on Pharmacoepidemiology & Therapeutic Risk Management Pre-conference educational session Thursday, October 23, 2014; 2:00-6:00pm Semi-automated,

More information

1201 Maryland Avenue SW, Suite 900, Washington, DC ,

1201 Maryland Avenue SW, Suite 900, Washington, DC , 1201 Maryland Avenue SW, Suite 900, Washington, DC 20024 202-962-9200, www.bio.org December 28, 2010 Dockets Management Branch (HFA-305) Food and Drug Administration 5600 Fishers Lane, Rm. 1061 Rockville,

More information

Draft Guidance for Industry, Clinical Laboratories, and FDA Staff. In Vitro Diagnostic Multivariate Index Assays

Draft Guidance for Industry, Clinical Laboratories, and FDA Staff. In Vitro Diagnostic Multivariate Index Assays Draft Guidance for Industry, Clinical Laboratories, and FDA Staff In Vitro Diagnostic Multivariate Index Assays DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Document

More information

Advance Topics in Pharmacoepidemiology. Risk Management. Conflict of Interest Declaration. Benefit Harm Profile?

Advance Topics in Pharmacoepidemiology. Risk Management. Conflict of Interest Declaration. Benefit Harm Profile? Advance Topics in Pharmacoepidemiology Risk Management 2012 Mid-Year ISPE Meeting Miami, April 21-23, 2012 Ariel E., Arias MD, PhD - Fac. Pharmacy; Université de Montréal - Biologics & Genetic Therapies

More information

August 25, Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm Rockville, MD 20852

August 25, Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm Rockville, MD 20852 August 25, 2014 Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm. 1061 Rockville, MD 20852 Re: FDA Docket No. FDA-2014-D-0758-0001 Draft Guidance for Industry on Distributing

More information

Re: Docket No. FDA-2018-D-1895: Indications and Usage Section of Labeling for Human Prescription Drug and Biological Products Content and Format

Re: Docket No. FDA-2018-D-1895: Indications and Usage Section of Labeling for Human Prescription Drug and Biological Products Content and Format September 7, 2018 Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm. 1061 Rockville, MD 20852 Re: Docket No. FDA-2018-D-1895: Indications and Usage Section of Labeling

More information

Potential Steps in Active Surveillance

Potential Steps in Active Surveillance Background The Sentinel System will augment FDA s postmarket safety assessment capabilities by enhancing its capacity to conduct active surveillance at the population level. Conducting active surveillance

More information

Adopted by the ENCePP Steering Group on 01/07/2016

Adopted by the ENCePP Steering Group on 01/07/2016 Doc.Ref. EMA/540136/2009 European Network of Centres for Pharmacoepidemiology and Pharmacovigilance Adopted by the ENCePP Steering Group on 01/07/2016 The European Network of Centres for Pharmacoepidemiology

More information

Guidance for Industry

Guidance for Industry Guidance for Industry Submission of Abbreviated Reports and Synopses in Support of Marketing Applications U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation

More information

Overview of FDA s Sentinel Initiative

Overview of FDA s Sentinel Initiative Overview of FDA s Sentinel Initiative Judy Racoosin, Sentinel Initiative Scientific Lead, Office of Medical Policy, Center for Drug Evaluation and Research, U.S. Food and Drug Administration September

More information

Guidance for Sponsors, Institutional Review Boards, Clinical Investigators and FDA Staff

Guidance for Sponsors, Institutional Review Boards, Clinical Investigators and FDA Staff Guidance for Sponsors, Institutional Review Boards, Clinical Investigators and FDA Staff Guidance on Informed Consent for In Vitro Diagnostic Device Studies Using Leftover Human Specimens that are Not

More information

Guidance for Clinical Investigators, Sponsors, and IRBs

Guidance for Clinical Investigators, Sponsors, and IRBs Guidance for Clinical Investigators, Sponsors, and IRBs Adverse Event Reporting to IRBs Improving Human Subject Protection u.s. Department of Health and Human Services Office of the Commissioner (OC) Center

More information

FDA Requirements 5/16/2011. FDA and new IND regs. Changes on the horizon: CIOMS IX ICH upcoming initiatives

FDA Requirements 5/16/2011. FDA and new IND regs. Changes on the horizon: CIOMS IX ICH upcoming initiatives Session Overview (1) Although the role of the Qualified Person responsible for Pharmacovigilance is mandated by the European legislation, the QPPV is held responsible for the establishment and the maintenance

More information

Guidance for Industry Pediatric Study Plans: Content of and Process for Submitting Initial Pediatric Study Plans and Amended Pediatric Study Plans

Guidance for Industry Pediatric Study Plans: Content of and Process for Submitting Initial Pediatric Study Plans and Amended Pediatric Study Plans Guidance for Industry Pediatric Study Plans: Content of and Process for Submitting Initial Pediatric Study Plans and Amended Pediatric Study Plans DRAFT GUIDANCE This guidance document is being distributed

More information

Nonproprietary Naming of Biological Products

Nonproprietary Naming of Biological Products Nonproprietary Naming of Biological Products Guidance for Industry DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Comments and suggestions regarding this draft document

More information

Compounding Animal Drugs From Bulk Drug Substances; Draft Guidance for Industry;

Compounding Animal Drugs From Bulk Drug Substances; Draft Guidance for Industry; This document is scheduled to be published in the Federal Register on 05/19/2015 and available online at http://federalregister.gov/a/2015-11982, and on FDsys.gov 4164-01-P DEPARTMENT OF HEALTH AND HUMAN

More information

Re: Docket No. FDA-2015-D-4562: Draft Guidance Safety Assessment for IND Safety Reporting

Re: Docket No. FDA-2015-D-4562: Draft Guidance Safety Assessment for IND Safety Reporting February 16, 2016 Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm. 1061 Rockville, MD 20852 Re: Docket No. FDA-2015-D-4562: Draft Guidance Safety Assessment for IND

More information

Report on the Performance of Drug and Biologics Firms in Conducting Postmarketing

Report on the Performance of Drug and Biologics Firms in Conducting Postmarketing This document is scheduled to be published in the Federal Register on 11/28/2016 and available online at https://federalregister.gov/d/2016-28442, and on FDsys.gov 4164-01-P DEPARTMENT OF HEALTH AND HUMAN

More information

Established Conditions: Reportable CMC Changes for Approved Drug and Biologic Products Guidance for Industry

Established Conditions: Reportable CMC Changes for Approved Drug and Biologic Products Guidance for Industry Established Conditions: Reportable CMC Changes for Approved Drug and Biologic Products Guidance for Industry DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Comments

More information

Draft Guidance for Industry on Direct-to-Consumer Television Advertisements--the Food and

Draft Guidance for Industry on Direct-to-Consumer Television Advertisements--the Food and This document is scheduled to be published in the Federal Register on 03/13/2012 and available online at http://federalregister.gov/a/2012-06040, and on FDsys.gov 4160-01-P DEPARTMENT OF HEALTH AND HUMAN

More information

Re: Docket No. FDA 2005-D-0339: Draft Guidance on Drug Safety Information FDA's Communication to the Public

Re: Docket No. FDA 2005-D-0339: Draft Guidance on Drug Safety Information FDA's Communication to the Public 1201 Maryland Avenue SW, Suite 900, Washington, DC 20024 202-962-9200, www.bio.org May 8, 2012 Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm. 1061 Rockville,

More information

STATEMENT SANDRA KWEDER, M.D DEPUTY DIRECTOR, OFFICE OF NEW DRUGS CENTER FOR DRUG EVALUATION AND RESEARCH U.S. FOOD AND DRUG ADMINISTRATION BEFORE THE

STATEMENT SANDRA KWEDER, M.D DEPUTY DIRECTOR, OFFICE OF NEW DRUGS CENTER FOR DRUG EVALUATION AND RESEARCH U.S. FOOD AND DRUG ADMINISTRATION BEFORE THE DEPARTMENT OF HEALTH & HUMAN SERVICES Public Health Service Food and Drug Administration Rockville MD 20857 STATEMENT OF SANDRA KWEDER, M.D DEPUTY DIRECTOR, OFFICE OF NEW DRUGS CENTER FOR DRUG EVALUATION

More information

Regulatory Aspects of Pharmacovigilance

Regulatory Aspects of Pharmacovigilance Regulatory Aspects of Pharmacovigilance Deirdre Mc Carthy Pia Caduff-Janosa Training Course Uppsala 2012 Agenda Risk based approach to spontaneous reporting (incl clinical trials) -> Pia Caduff-Janosa

More information

The Right Data for the Right Questions:

The Right Data for the Right Questions: The Right Data for the Right Questions: Evidentiary Needs as a Guide to Data Source Selection David Thompson, PhD Senior Vice President Louise Parmenter, PhD Global Head of Operations Real-World & Late

More information

USE OF PRIMARY CARE DATABASES IN EPIDEMIOLOGIC RESEARCH

USE OF PRIMARY CARE DATABASES IN EPIDEMIOLOGIC RESEARCH USE OF PRIMARY CARE DATABASES IN EPIDEMIOLOGIC RESEARCH Luis Alberto García Rodríguez Spanish Centre for Pharmacoepidemiologic Research (CEIFE), Madrid, Spain Index The structure to successfully perform

More information

What s New in GCP? FDA Clarifies, Expands Safety Reporting Guidance

What s New in GCP? FDA Clarifies, Expands Safety Reporting Guidance Vol. 9, No. 2, February 2013 Happy Trials to You What s New in GCP? FDA Clarifies, Expands Safety Reporting Guidance Reprinted from the Guide to Good Clinical Practice with permission of Thompson Publishing

More information

Draft Guidance for Industry and Food and Drug Administration Staff

Draft Guidance for Industry and Food and Drug Administration Staff Draft Guidance for Industry and Food and Drug Administration Staff Investigational Device Exemptions (IDE) for Early Feasibility Medical Device Clinical Studies, Including Certain First in Human (FIH)

More information

Guidelines: c. Providing the investigational drug for the requested use will not interfere with the initiation, conduct, or completion of clinical

Guidelines: c. Providing the investigational drug for the requested use will not interfere with the initiation, conduct, or completion of clinical Guidelines for the Submission of an Expanded Access IND to Permit Diagnosis, Monitoring or Treatment of Intermediate-size Patient Populations with an Investigational Drug or a REMS-restricted, Approved

More information

Appendix 3: Another look at the US IND annual report

Appendix 3: Another look at the US IND annual report Appendix 3: Another look at the US IND annual report As the Development Safety Update Report (DSUR) came into effect in late 2011, Chapter 2 (Pharmacovigilance Medical Writing for Clinical Trials) of the

More information

Re: Comments on January 2017 Draft Guidance: Considerations in Demonstrating Interchangeability with a Reference Product (Docket No.

Re: Comments on January 2017 Draft Guidance: Considerations in Demonstrating Interchangeability with a Reference Product (Docket No. 800 17th Street, NW Suite 1100, Washington, DC 20006 May 1, 2017 Dr. Stephen Ostroff Acting Commissioner Food and Drug Administration 10903 New Hampshire Avenue Silver Spring, MD 20993 Re: Comments on

More information

Guidance for IRBs, Clinical Investigators and Sponsors

Guidance for IRBs, Clinical Investigators and Sponsors Guidance for IRBs, Clinical Investigators and Sponsors IRB Responsibilities for Reviewing the Qualifications of Investigators, Adequacy of Research Sites, and the Determination of Whether an IND/IDE is

More information

Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, rm Rockville, MD 20852

Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, rm Rockville, MD 20852 Reference No.: FDAA10017 Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, rm. 1061 Rockville, MD 20852 VIA WEB SUBJECT: Approval Pathway for Biosimilar and Interchangeable

More information

Health Canada s NHP Vigilance Activities

Health Canada s NHP Vigilance Activities Health Canada s NHP Vigilance Activities Canadian Health Food Association (CHFA) East October 3, 2013 Scott Sawler Director General Marketed Health Products Directorate Overview NHP Program & MHPD Activities

More information

Question Yes No. 1. Is the study interventional (a clinical trial)? Study Type data element is Interventional

Question Yes No. 1. Is the study interventional (a clinical trial)? Study Type data element is Interventional Checklist for Evaluating Whether a Clinical Trial or Study is an Applicable Clinical Trial (ACT) Under 42 CFR 11.22(b) for Clinical Trials Initiated on or After January 18, 2017 1 (NOT FOR SUBMISSION 2

More information

Overcoming Statistical Challenges to the Reuse of Data within the Mini-Sentinel Distributed Database

Overcoming Statistical Challenges to the Reuse of Data within the Mini-Sentinel Distributed Database September 5, 2012 September 5, 2012 Meeting Summary Overcoming Statistical Challenges to the Reuse of Data within the Mini-Sentinel Distributed Database With passage of the Food and Drug Administration

More information

Post-marketing Drug Safety Surveillance: Pharmacovigilance in FDA/CDER

Post-marketing Drug Safety Surveillance: Pharmacovigilance in FDA/CDER Post-marketing Drug Safety Surveillance: Pharmacovigilance in FDA/CDER CSRC/DIA Think Tank June 3, 2016 Oanh Dang, PharmD, BCPS Division of Pharmacovigilance Safety Evaluator Office of Surveillance and

More information

Guidance for Industry Q3B(R2) Impurities in New Drug Products

Guidance for Industry Q3B(R2) Impurities in New Drug Products Guidance for Industry Q3B(R2) Impurities in New Drug Products U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics

More information

Statement on Regulatory Systems to Improve Pharmaceutical Safety

Statement on Regulatory Systems to Improve Pharmaceutical Safety International Society for Pharmacoepidemiology (ISPE) Statement on Regulatory Systems to Improve Pharmaceutical Safety INTRODUCTION The public, some researchers, individual regulators and policymakers

More information

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) European Medicines Agency Pre-authorisation Evaluation of Medicines for Human Use London, 22 February 2006 EMEA/CHMP/BMWP/42832/2005 COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON SIMILAR

More information

De Novo Classification Process (Evaluation of Automatic Class III Designation) Guidance for Industry and Food and Drug Administration Staff

De Novo Classification Process (Evaluation of Automatic Class III Designation) Guidance for Industry and Food and Drug Administration Staff De Novo Classification Process (Evaluation of Automatic Class III Designation) Guidance for Industry and Food and Drug Administration Staff Document issued on October 30, 2017. The draft of this document

More information

Disclosures. Laboratory Stakeholders. IVD vs. LDT. FDA Regulation of Laboratory Developed Tests 10/2/2015. FDA Regulation of LDTs

Disclosures. Laboratory Stakeholders. IVD vs. LDT. FDA Regulation of Laboratory Developed Tests 10/2/2015. FDA Regulation of LDTs Disclosures FDA Regulation of Laboratory Developed Tests Beaumont Health System, 24 th Annual Symposium on Molecular Pathology September 16, 2015 Roger D. Klein, MD JD Director, Molecular Pathology Clinical

More information

FDA from a Former FDAer: Secrets and insights into regulatory review and drug development

FDA from a Former FDAer: Secrets and insights into regulatory review and drug development FDA from a Former FDAer: Secrets and insights into regulatory review and drug development Andrew E. Mulberg, MD, FAAP Vice-President, Global Regulatory Affairs; Former Division Deputy, DGIEP, U.S. FDA

More information

USE OF INVESTIGATIONAL DRUGS OR BIOLOGICS IN HUMAN SUBJECTS RESEARCH

USE OF INVESTIGATIONAL DRUGS OR BIOLOGICS IN HUMAN SUBJECTS RESEARCH Policy P.11 Written By: B. Laurel Elder Created: June 3, 2013 Edited Replaces 10-9-2009 Version P.11.4 USE OF INVESTIGATIONAL DRUGS OR BIOLOGICS IN HUMAN SUBJECTS RESEARCH A. Procedure Contents this procedure

More information

Pragmatic Clinical Trials for Regulatory Decisions

Pragmatic Clinical Trials for Regulatory Decisions Pragmatic Clinical Trials for Regulatory Decisions Jacqueline Corrigan-Curay, MD JD Office of Medical Policy Center for Drug Evaluation and Research FDA May 16, 2018 Pragmatic Clinical Trials Pragmatic

More information

Guidance for Industry Reference Product Exclusivity for Biological Products Filed Under Section 351(a) of the PHS Act

Guidance for Industry Reference Product Exclusivity for Biological Products Filed Under Section 351(a) of the PHS Act Guidance for Industry Reference Product Exclusivity for Biological Products Filed Under Section 351(a) of the PHS Act DRAFT GUIDANCE This guidance document is being distributed for comment purposes only.

More information

Docket #: FDA-2018-D-3268

Docket #: FDA-2018-D-3268 Subject: Comment on FDA Draft Guidance for Industry Titled Rare Diseases: Early Drug Development and the Role of Pre-Investigational New Drug Application Meetings Docket #: FDA-2018-D-3268 ARM is an international

More information

Disproportionality Analysis and Its Application to Spontaneously Reported Adverse Events in Pharmacovigilance

Disproportionality Analysis and Its Application to Spontaneously Reported Adverse Events in Pharmacovigilance WHITE PAPER Disproportionality Analysis and Its Application to Spontaneously Reported Adverse Events in Pharmacovigilance Richard C. Zink, SAS, Cary, NC Table of Contents Introduction... 1 Spontaneously

More information

THE FDA, THE DRUG APPROVAL PROCESS, AND THE PATIENT VOICE

THE FDA, THE DRUG APPROVAL PROCESS, AND THE PATIENT VOICE THE FDA, THE DRUG APPROVAL PROCESS, AND THE PATIENT VOICE Ali Mohamadi, M.D. Senior Director, Global Regulatory Patient Engagement and Policy BioMarin Pharmaceutical 30-July-2016 Goals of Today s Presentation

More information

Page2 Docket No. FDA-2013-N-0745 November 20, 2013

Page2 Docket No. FDA-2013-N-0745 November 20, 2013 Via Electronic Submission Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm. 1061 Rockville, MD 20852 Re: Food and Drug Administration Safety and Innovation Act

More information

Subpart H [Reserved] Subpart I Expanded Access to Investigational Drugs for Treatment. 21 CFR Ch. I ( Edition)

Subpart H [Reserved] Subpart I Expanded Access to Investigational Drugs for Treatment. 21 CFR Ch. I ( Edition) 312.300 (c) Disposition of unused drug. The person who ships the drug under paragraph (a) of this section shall assure the return of all unused supplies of the drug from individual investigators whenever

More information

Pharmacovigilance Practice in Pharmaceutical Industry. From Adverse Event Collection Risk Management

Pharmacovigilance Practice in Pharmaceutical Industry. From Adverse Event Collection Risk Management Pharmacovigilance Practice in Pharmaceutical Industry From Adverse Event Collection Risk Management Hani Mickail, MD Head Global Clinical Safety Operations - Novartis Pharmacovigilance Workshop 23-24 October

More information

Vaccine Safety Monitoring, Reporting and Analysis. GBC 2017, Yun Chon, Ph. D

Vaccine Safety Monitoring, Reporting and Analysis. GBC 2017, Yun Chon, Ph. D Vaccine Safety Monitoring, Reporting and Analysis GBC 2017, Yun Chon, Ph. D Disclaimer This presentation is based on my own opinion and is not representing the opinion of International Vaccine Institute.

More information

Regulatory Statistical Perspectives on Safety Issues in Drug Development

Regulatory Statistical Perspectives on Safety Issues in Drug Development Regulatory Statistical Perspectives on Safety Issues in Drug Development C. George Rochester, Ph.D. Lead Statistician for Drug Safety Evaluation Food & Drug Administration, Rockville, MD, USA RochesterG@cder.fda.gov

More information

Office for Human Subject Protection. University of Rochester

Office for Human Subject Protection. University of Rochester POLICY 1. Purpose Outline the responsibilities and regulatory requirements when conducting human subject research that involves the use of drugs, agents, biological products, or nutritional products (e.g.,

More information

FDA Public Hearing: Approval Pathway for Biosimilar. Products. November 2-3, 2010

FDA Public Hearing: Approval Pathway for Biosimilar. Products. November 2-3, 2010 FDA Public Hearing: Approval Pathway for Biosimilar and Interchangeable Biological Products November 2-3, 2010 1 The Biotechnology Industry Organization Over 1,100 members, including biotechnology companies,

More information

GUIDANCE FOR INDUSTRY. Questions and Answers

GUIDANCE FOR INDUSTRY. Questions and Answers GUIDANCE FOR INDUSTRY Prescription Drug Marketing Act (PDMA) Requirements Questions and Answers U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and

More information

May 9, Meeting Summary. Facilitating Antibacterial Drug Development

May 9, Meeting Summary. Facilitating Antibacterial Drug Development May 9, 2012 Meeting Summary Facilitating Antibacterial Drug Development Origins of the Current Public Health Crisis of Antibacterial Resistance Antibacterial drugs play a critical role in the ability to

More information

Compassionate Use Navigator Information for Physicians

Compassionate Use Navigator Information for Physicians Compassionate Use Navigator Information for Physicians Contact: Elena Gerasimov, Program Director, Elena@kidsvcancer.org. As a physician, you must have wished there would be more treatment options for

More information

Expanded Access. to Investigational Drugs & Biologics. for Treatment Use

Expanded Access. to Investigational Drugs & Biologics. for Treatment Use SJMHS Research Compliance Office Guidance Document Expanded Access to Investigational Drugs & Biologics for Treatment Use May 2015 1 Expanded Access to Investigational Drugs & Biologics for Treatment Use

More information

GUIDELINE REGARDING COLLECTION, VERIFICATION, AND SUBMISSION OF THE REPORTS OF ADVERSE EVENTS / REACTIONS OCCURRING IN CLINICAL DRUG TRIALS

GUIDELINE REGARDING COLLECTION, VERIFICATION, AND SUBMISSION OF THE REPORTS OF ADVERSE EVENTS / REACTIONS OCCURRING IN CLINICAL DRUG TRIALS 1. PURPOSE This guideline is about the collection, verification, and submission of the reports of adverse events / reactions occurring in clinical drug trials, and code breaking methods. 2. DEFINITIONS

More information

2/11/2014. Key Sources of Uncertainty in the Assessment of Benefits and Risks of Pharmaceuticals and Associated Challenges. Disclaimer.

2/11/2014. Key Sources of Uncertainty in the Assessment of Benefits and Risks of Pharmaceuticals and Associated Challenges. Disclaimer. Key Sources of Uncertainty in the Assessment of Benefits and Risks of Pharmaceuticals and Associated Challenges Institute of Medicine Workshop: Characterizing and Communicating Uncertainty in the Assessment

More information

Comments on Draft Guidance for Industry Considerations in Demonstrating Interchangeability with a Reference Product [Docket No.

Comments on Draft Guidance for Industry Considerations in Demonstrating Interchangeability with a Reference Product [Docket No. Pfizer Inc 235 East 42 nd Street New York, NY 10017-5755 24 March 2017 By Electronic Submission. Division of Dockets Management (HFA-305), Food and Drug Administration, 5630 Fishers Lane, rm. 1061, Rockville,

More information

GUIDANCE FOR SPONSORS, INDUSTRY, RESEARCHERS, INVESTIGATORS, AND FOOD AND DRUG ADMINISTRATION STAFF

GUIDANCE FOR SPONSORS, INDUSTRY, RESEARCHERS, INVESTIGATORS, AND FOOD AND DRUG ADMINISTRATION STAFF GUIDANCE FOR SPONSORS, INDUSTRY, RESEARCHERS, INVESTIGATORS, AND FOOD AND DRUG ADMINISTRATION STAFF Certifications To Accompany Drug, Biological Product, and Device Applications/Submissions:Compliance

More information

Guidance for Industry Compounding Animal Drugs from Bulk Drug Substances

Guidance for Industry Compounding Animal Drugs from Bulk Drug Substances #230 Guidance for Industry Compounding Animal Drugs from Bulk Drug Substances DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Comments and suggestions regarding this

More information

Re: Docket No. 2006D-0347, Federal Register: July 26, 2007 (Volume 72, Pages )

Re: Docket No. 2006D-0347, Federal Register: July 26, 2007 (Volume 72, Pages ) 1201 Maryland Avenue SW, Ste. 900 Washington, DC 20024 August 27, 2007 BY ELECTRONIC DELIVERY Division of Dockets Management (HFA-305) Food and Drug Administration 56350 Fishers Lane Room 1061 Rockville,

More information

Use of Real-World Evidence to Support Regulatory Decision-Making for Medical Devices

Use of Real-World Evidence to Support Regulatory Decision-Making for Medical Devices 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 Use of Real-World Evidence to Support Regulatory Decision-Making for Medical Devices Draft Guidance

More information

Surveillance Part 2. Marcia A. Testa, MPH, PhD Harvard School of Public Health

Surveillance Part 2. Marcia A. Testa, MPH, PhD Harvard School of Public Health Surveillance Part 2 Marcia A. Testa, MPH, PhD Harvard School of Public Health Periodic Population-based Surveys Used for surveillance if surveys are repeated on a regular basis Careful attention to the

More information

Improving Risk Management and Drug Safety. Janet Woodcock, MD Director Center for Drug Evaluation and Research U.S. Food and Drug Administration

Improving Risk Management and Drug Safety. Janet Woodcock, MD Director Center for Drug Evaluation and Research U.S. Food and Drug Administration Improving Risk Management and Drug Safety Janet Woodcock, MD Director Center for Drug Evaluation and Research U.S. Food and Drug Administration November 9, 2010 TOPICS Sentinel Initiative Risk Evaluation

More information

Structure and Mandate of FDA

Structure and Mandate of FDA Structure and Mandate of FDA Leonard Sacks, M.D. Office of Medical Policy Center for Drug Evaluation and Research FDA FDA Clinical Investigator Training Course November 13, 2018 Mission of regulatory agencies

More information

SUBJECT: Public Meeting on Expansion of the Clinical Trial Registry and Results Data Bank [Docket No. NIH ]

SUBJECT: Public Meeting on Expansion of the Clinical Trial Registry and Results Data Bank [Docket No. NIH ] Date: June 22, 2009 Reference No.: FDAA09008 VIA WEB & USPS National Institute of Health National Library of Medicine 6705 Rockledge Drive, Suite 301 Bethesda, MD 20892 SUBJECT: Public Meeting on Expansion

More information

New and Revised Draft Q&As on Biosimilar Development and the BPCI Act (Revision 2)

New and Revised Draft Q&As on Biosimilar Development and the BPCI Act (Revision 2) New and Revised Draft Q&As on Biosimilar Development and the BPCI Act (Revision 2) Guidance for Industry DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Comments and

More information

The Future of Drug Safety

The Future of Drug Safety The Future of Drug Safety IOM Committee on the Assessment of the US Drug Safety System R. Alta Charo Warren P. Knowles Professor of Law & Bioethics University of Wisconsin Charge to the Committee Examine

More information

FDA N-5319 VIA ELECTRONIC SUBMISSION. January 10, 2018

FDA N-5319 VIA ELECTRONIC SUBMISSION. January 10, 2018 1227 25th St. NW #700 Washington, DC 20037 combinationproducts.com 202.861.4199 VIA ELECTRONIC SUBMISSION January 10, 2018 Division of Dockets Management (HFA-305) Food and Drug Administration 5630 Fishers

More information

FDA Decisions for Investigational Device Exemption (IDE) Clinical Investigations

FDA Decisions for Investigational Device Exemption (IDE) Clinical Investigations Draft Guidance for Industry, Clinical Investigators, Institutional Review Boards, and Food and Drug Administration Staff FDA Decisions for Investigational Device Exemption (IDE) Clinical Investigations

More information

Guidance for Industry

Guidance for Industry Reprinted from FDA s website by Guidance for Industry Scientific Considerations in Demonstrating Biosimilarity to a Reference Product DRAFT GUIDANCE This guidance document is being distributed for comment

More information

22 January Division of Dockets Management (HFA 305) Food and Drug Administration 5630 Fishers Lane, rm Rockville, MD 20852

22 January Division of Dockets Management (HFA 305) Food and Drug Administration 5630 Fishers Lane, rm Rockville, MD 20852 22 January 2009 Division of Dockets Management (HFA 305) Food and Drug Administration 5630 Fishers Lane, rm. 1061 Rockville, MD 20852 SUBMISSION OF COMMENTS, DOCKET NO. FDA-2008-D-0559 Dear Sir or Madam:

More information

Re: Docket Number FDA-2017-D-0154 Considerations in Demonstrating Interchangeability With a Reference Product; Guidance for Industry, Draft Guidance

Re: Docket Number FDA-2017-D-0154 Considerations in Demonstrating Interchangeability With a Reference Product; Guidance for Industry, Draft Guidance PO Box 3691 Arlington, VA 22203 (703) 971-1700 May 22, 2017 Division of Dockets Management (HFA-305) Food and Drug Administration Department of Health and Human Services 5630 Fishers Lane, Room 1061 Rockville,

More information

Guidelines for Detecting & Reporting Adverse Drug Reactions

Guidelines for Detecting & Reporting Adverse Drug Reactions Guidelines for Detecting & Reporting Adverse Drug Reactions For Healthcare Professionals Rational Drug Use and Pharmacovigilance Department- JFDA (2014) Guidelines for Detecting & Reporting Adverse Drug

More information

Evaluation of existing methods for safety signal identification for the Sentinel Initiative

Evaluation of existing methods for safety signal identification for the Sentinel Initiative Evaluation of existing methods for safety signal identification for the Sentinel Initiative Jennifer C. Nelson Biostatistics Unit Group Health Research Institute Department of Biostatistics University

More information

Expiration Dating of Unit-Dose Repackaged Solid Oral Dosage Form Drug Products Guidance for Industry

Expiration Dating of Unit-Dose Repackaged Solid Oral Dosage Form Drug Products Guidance for Industry Expiration Dating of Unit-Dose Repackaged Solid Oral Dosage Form Drug Products Guidance for Industry DRAFT GUIDANCE This guidance document is being distributed for comment purposes only. Comments and suggestions

More information

Drug Safety and FDA Revitalization Legislation

Drug Safety and FDA Revitalization Legislation Drug Safety and FDA Revitalization Legislation Dan Kracov August 23, 2007 FDA Regulatory and Compliance Symposium House and Senate FDA Revitalization Bills Senate: S. 1082, Food and Drug Administration

More information

Topics. Safety First Initiative. Drug Safety Communications DRUG SAFETY INITIATIVES AT THE FOOD AND DRUG ADMINISTRATION

Topics. Safety First Initiative. Drug Safety Communications DRUG SAFETY INITIATIVES AT THE FOOD AND DRUG ADMINISTRATION DRUG SAFETY INITIATIVES AT THE FOOD AND DRUG ADMINISTRATION Presentation to the FDA/Industry Conference sponsored by the School of Pharmacy at Temple University May 6, 2008 Topics Safety First initiative

More information